Try a new search

Format these results:

Searched for:

person:nixonr01 or ginsbs01 or levye01 or mathep01 or ohnom01 or raom01 or scharh01 or yangd02 or yuana01

Total Results:

1130


Endocrine Insights into the Pathophysiology of Autism Spectrum Disorder

Wilson, Hayley A; Creighton, Carolyn; Scharfman, Helen; Choleris, Elena; MacLusky, Neil J
Autism spectrum disorder (ASD) is a class of neurodevelopmental disorders that affects males more frequently than females. Numerous genetic and environmental risk factors have been suggested to contribute to the development of ASD. However, no one factor can adequately explain either the frequency of the disorder or the male bias in its prevalence. Gonadal, thyroid, and glucocorticoid hormones all contribute to normal development of the brain, hence perturbations in either their patterns of secretion or their actions may constitute risk factors for ASD. Environmental factors may contribute to ASD etiology by influencing the development of neuroendocrine and neuroimmune systems during early life. Emerging evidence suggests that the placenta may be particularly important as a mediator of the actions of environmental and endocrine risk factors on the developing brain, with the male being particularly sensitive to these effects. Understanding how various risk factors integrate to influence neural development may facilitate a clearer understanding of the etiology of ASD.
PMID: 32912048
ISSN: 1089-4098
CID: 4598212

Extracellular vesicles: where the amyloid precursor protein carboxyl-terminal fragments accumulate and amyloid-β oligomerizes

Pérez-González, Rocío; Kim, Yohan; Miller, Chelsea; Pacheco-Quinto, Javier; Eckman, Elizabeth A; Levy, Efrat
Pleiotropic roles are proposed for brain extracellular vesicles (EVs) in the development of Alzheimer's disease (AD). Our previous studies have suggested a beneficial role for EVs in AD, where the endosomal system in vulnerable neurons is compromised, contributing to the removal of accumulated material from neurons. However, the involvement of EVs in propagating AD amyloidosis throughout the brain has been considered because the amyloid-β precursor protein (APP), APP metabolites, and key APP cleaving enzymes were identified in association with EVs. Here, we undertook to determine whether the secretase machinery is actively processing APP in EVs isolated from the brains of wild-type and APP overexpressing Tg2576 mice. We found that full-length APP is cleaved in EVs incubated in the absence of cells. The resulting metabolites, both α- and β-APP carboxyl-terminal fragments and APP intracellular domain accumulate in EVs over time and amyloid-β dimerizes. Thus, EVs contribute to the removal from neurons and transport of APP-derived neurotoxic peptides. While this is potentially a venue for propagation of the pathology throughout the brain, it may contribute to efficient removal of neurotoxic peptides from the brain.
PMID: 32772431
ISSN: 1530-6860
CID: 4560212

Neurofilaments: neurobiological foundations for biomarker applications

Gafson, Arie R; Barthélemy, Nicolas R; Bomont, Pascale; Carare, Roxana O; Durham, Heather D; Julien, Jean-Pierre; Kuhle, Jens; Leppert, David; Nixon, Ralph A; Weller, Roy O; Zetterberg, Henrik; Matthews, Paul M
Interest in neurofilaments has risen sharply in recent years with recognition of their potential as biomarkers of brain injury or neurodegeneration in CSF and blood. This is in the context of a growing appreciation for the complexity of the neurobiology of neurofilaments, new recognition of specialized roles for neurofilaments in synapses and a developing understanding of mechanisms responsible for their turnover. Here we will review the neurobiology of neurofilament proteins, describing current understanding of their structure and function, including recently discovered evidence for their roles in synapses. We will explore emerging understanding of the mechanisms of neurofilament degradation and clearance and review new methods for future elucidation of the kinetics of their turnover in humans. Primary roles of neurofilaments in the pathogenesis of human diseases will be described. With this background, we then will review critically evidence supporting use of neurofilament concentration measures as biomarkers of neuronal injury or degeneration. Finally, we will reflect on major challenges for studies of the neurobiology of intermediate filaments with specific attention to identifying what needs to be learned for more precise use and confident interpretation of neurofilament measures as biomarkers of neurodegeneration.
PMID: 32408345
ISSN: 1460-2156
CID: 4438212

Translational neurophysiological biomarkers of N-methyl-d-aspartate receptor dysfunction in serine racemase knockout mice

Balla, Andrea; Ginsberg, Stephen D; Abbas, Atheir I; Sershen, Henry; Javitt, Daniel C
Alterations in glutamatergic function are well established in schizophrenia (Sz), but new treatment development is hampered by the lack of translational pathophysiological and target engagement biomarkers as well as by the lack of animal models that recapitulate the pathophysiological features of Sz. Here, we evaluated the rodent auditory steady state response (ASSR) and long-latency auditory event-related potential (aERP) as potential translational markers. These biomarkers were assessed for their sensitivity to both the N-methyl-d-aspartate receptor (NMDAR) antagonist phencyclidine (PCP) and to knock-out (KO) of Serine Racemase (SR), which is known to lead to Sz-like alterations in function of parvalbumin (PV)-type cortical interneurons. PCP led to significant increases of ASSR that were further increased in SRKO-/-, consistent with PV interneuron effects. Similar effects were observed in mice with selective NMDAR KO on PV interneurons. By contrast, PCP but not SRKO reduced the amplitude of the rodent analog of the human N1 potential. Overall, these findings support use of rodent ASSR and long-latency aERP, along with previously described measures such as mismatch negativity (MMN), as translational biomarkers, and support SRKO mice as a potential rodent model for PV interneuron dysfunction in Sz.
PMCID:8301266
PMID: 34308374
ISSN: 2666-1446
CID: 4965432

The aging lysosome: An essential catalyst for late-onset neurodegenerative diseases

Nixon, Ralph A
Lysosomes figure prominently in theories of aging as the proteolytic system most responsible for eliminating growing burdens of damaged proteins and organelles in aging neurons and other long lived cells. Newer evidence shows that diverse experimental measures known to extend lifespan in invertebrate aging models share the property of boosting lysosomal clearance of substrates through the autophagy pathway. Maintaining an optimal level of lysosome acidification is particularly crucial for these anti-aging effects. The exceptional dependence of neurons on fully functional lysosomes is reflected by the phenotypes seen in congenital lysosomal storage disorders, which commonly present as severe neurodevelopmental or neurodegenerative conditions even though lysosomal deficits are systemic. Similar connections are now being appreciated between risk for late age-onset neurodegenerative disorders and primary lysosomal deficits. In diseases such as Alzheimer's and Parkinson's, as in aging alone, primary lysosome dysfunction due to acidification impairment is emerging as a frequent theme, supported by the growing list of familial neurodegenerative disorders that involve primary vATPase dysfunction. The additional cellular roles played by intraluminal pH in sensing nutrient and stress and modulating cellular signaling have further expanded the possible ways that lysosomal pH dysregulation in aging and disease can disrupt neuronal function. Here, we consider the impact of cellular aging on lysosomes and how these changes may create the tipping point for disease emergence in major late-age onset neurodegenerative disorders.
PMID: 32416272
ISSN: 1878-1454
CID: 4438412

β2-adrenergic Agonists Rescue Lysosome Acidification and Function in PSEN1 Deficiency by Reversing Defective ER-to-lysosome Delivery of ClC-7

Lee, Ju-Hyun; Wolfe, Devin M; Darji, Sandipkumar; McBrayer, Mary Kate; Colacurcio, Daniel J; Kumar, Asok; Stavrides, Philip; Mohan, Panaiyur S; Nixon, Ralph A
Lysosomal dysfunction is considered pathogenic in Alzheimer disease (AD). Loss of presenilin-1 (PSEN1) function causing AD impedes acidification via defective vacuolar ATPase (vATPase) V0a1 subunit delivery to lysosomes. We report that isoproterenol (ISO) and related β2-adrenergic agonists reacidify lysosomes in PSEN1 Knock out (KO) cells and fibroblasts from PSEN1 familial AD patients, which restores lysosomal proteolysis, calcium homeostasis, and normal autophagy flux. We identify a novel rescue mechanism involving Portein Kinase A (PKA)-mediated facilitation of chloride channel-7 (ClC-7) delivery to lysosomes which reverses markedly lowered chloride (Cl-) content in PSEN1 KO lysosomes. Notably, PSEN1 loss of function impedes Endoplasmic Reticulum (ER)-to-lysosome delivery of ClC-7. Transcriptomics of PSEN1-deficient cells reveals strongly downregulated ER-to-lysosome transport pathways and reversibility by ISO, thus accounting for lysosomal Cl- deficits that compound pH elevation due to deficient vATPase and its rescue by β2-adrenergic agonists. Our findings uncover a broadened PSEN1 role in lysosomal ion homeostasis and novel pH modulation of lysosomes through β2-adrenergic regulation of ClC-7, which can potentially be modulated therapeutically.
PMID: 32105735
ISSN: 1089-8638
CID: 4394692

Type I interferon response drives neuroinflammation and synapse loss in Alzheimer disease

Roy, Ethan R; Wang, Baiping; Wan, Ying-Wooi; Chiu, Gabriel; Cole, Allysa; Yin, Zhuoran; Propson, Nicholas E; Xu, Yin; Jankowsky, Joanna L; Liu, Zhandong; Lee, Virginia M-Y; Trojanowski, John Q; Ginsberg, Stephen D; Butovsky, Oleg; Zheng, Hui; Cao, Wei
Type I interferon (IFN) is a key cytokine that curbs viral infection and cell malignancy. Previously, we demonstrated a potent IFN immunogenicity of nucleic acid-containing (NA-containing) amyloid fibrils in the periphery. Here, we investigated whether IFN is associated with β-amyloidosis inside the brain and contributes to neuropathology. An IFN-stimulated gene (ISG) signature was detected in the brains of multiple murine Alzheimer disease (AD) models, a phenomenon also observed in WT mouse brain challenged with generic NA-containing amyloid fibrils. In vitro, microglia innately responded to NA-containing amyloid fibrils. In AD models, activated ISG-expressing microglia exclusively surrounded NA+ amyloid β plaques, which accumulated in an age-dependent manner. Brain administration of rIFN-β resulted in microglial activation and complement C3-dependent synapse elimination in vivo. Conversely, selective IFN receptor blockade effectively diminished the ongoing microgliosis and synapse loss in AD models. Moreover, we detected activated ISG-expressing microglia enveloping NA-containing neuritic plaques in postmortem brains of patients with AD. Gene expression interrogation revealed that IFN pathway was grossly upregulated in clinical AD and significantly correlated with disease severity and complement activation. Therefore, IFN constitutes a pivotal element within the neuroinflammatory network of AD and critically contributes to neuropathogenic processes.
PMCID:7108898
PMID: 31917687
ISSN: 1558-8238
CID: 4386212

The epichaperome is a mediator of toxic hippocampal stress and leads to protein connectivity-based dysfunction

Inda, Maria Carmen; Joshi, Suhasini; Wang, Tai; Bolaender, Alexander; Gandu, Srinivasa; Koren Iii, John; Che, Alicia Yue; Taldone, Tony; Yan, Pengrong; Sun, Weilin; Uddin, Mohammad; Panchal, Palak; Riolo, Matthew; Shah, Smit; Barlas, Afsar; Xu, Ke; Chan, Lon Yin L; Gruzinova, Alexandra; Kishinevsky, Sarah; Studer, Lorenz; Fossati, Valentina; Noggle, Scott A; White, Julie R; de Stanchina, Elisa; Sequeira, Sonia; Anthoney, Kyle H; Steele, John W; Manova-Todorova, Katia; Patil, Sujata; Dunphy, Mark P; Pillarsetty, NagaVaraKishore; Pereira, Ana C; Erdjument-Bromage, Hediye; Neubert, Thomas A; Rodina, Anna; Ginsberg, Stephen D; De Marco Garcia, Natalia; Luo, Wenjie; Chiosis, Gabriela
Optimal functioning of neuronal networks is critical to the complex cognitive processes of memory and executive function that deteriorate in Alzheimer's disease (AD). Here we use cellular and animal models as well as human biospecimens to show that AD-related stressors mediate global disturbances in dynamic intra- and inter-neuronal networks through pathologic rewiring of the chaperome system into epichaperomes. These structures provide the backbone upon which proteome-wide connectivity, and in turn, protein networks become disturbed and ultimately dysfunctional. We introduce the term protein connectivity-based dysfunction (PCBD) to define this mechanism. Among most sensitive to PCBD are pathways with key roles in synaptic plasticity. We show at cellular and target organ levels that network connectivity and functional imbalances revert to normal levels upon epichaperome inhibition. In conclusion, we provide proof-of-principle to propose AD is a PCBDopathy, a disease of proteome-wide connectivity defects mediated by maladaptive epichaperomes.
PMID: 31949159
ISSN: 2041-1723
CID: 4264582

Fixation protocols for neurohistology: neurons to genes

Mufson, E J; Perez, S E; Kelley, C M; Alldred, M J; Ginsberg, S D
Since ancient times, tissue fixation for neurohistology has been an evolving area of research. Alcohol fixation first made possible examination of brain tissue specimens. In the late 1800s formaldehyde was introduced as a cross-linking fixative, which enabled histological advances. Various aldehyde solutions remain the staple for neurohistology in the neuroscience community including formalin, paraformaldehyde, glutaraldehyde, and combinations of these fixatives. A 4% paraformaldehyde solution is commonly used for numerous cytochemical procedures including tract tracing, immunohistochemistry, and in situ hybridization. A glutaraldehyde-paraformaldehyde solution is the fixative of choice for ultrastructural investigations using the electron microscope. With the advent of modern molecular and cellular biological techniques, it is now possible to isolate and study genomic DNA, RNA species, and proteins from microdissected tissue sources. Similar to light and electron microscopy, alcohol and aldehyde tissue fixation are compatible with preserving RNA integrity for regional and single cell gene array experiments using immersion- and perfusion-fixed tissue from a myriad of brain tissue sources including humans and relevant animal and cellular models.
Copyright
EMBASE:632602397
ISSN: 1940-6045
CID: 4571842

Genes Bound by ΔFosB in Different Conditions With Recurrent Seizures Regulate Similar Neuronal Functions

Stephens, Gabriel S; Fu, Chia-Hsuan; St Romain, Corey P; Zheng, Yi; Botterill, Justin J; Scharfman, Helen E; Liu, Yin; Chin, Jeannie
Seizure incidence is increased in Alzheimer's disease (AD) patients and mouse models, and treatment with the antiseizure drug levetiracetam improves cognition. We reported that one mechanism by which seizures can exert persistent effects on cognition is through accumulation of ΔFosB, a transcription factor with a long half-life. Even the infrequent seizures that spontaneously occur in transgenic mice expressing human amyloid precursor protein (APP) lead to persistent increases in ΔFosB in the hippocampus, similar to what we observed in patients with AD or temporal lobe epilepsy. ΔFosB epigenetically regulates expression of target genes, however, whether ΔFosB targets the same genes when induced by seizures in different neurological conditions is not clear. We performed ChIP-sequencing to assess the repertoire of ΔFosB target genes in APP mice and in pilocarpine-treated wildtype mice (Pilo mice), a pharmacological model of epilepsy. These mouse models allowed us to compare AD, in which seizures occur in the context of high levels of amyloid beta, and epilepsy, in which recurrent seizures occur without AD-specific pathophysiology. Network profiling of genes bound by ΔFosB in APP mice, Pilo mice, and respective control mice revealed that functional domains modulated by ΔFosB in the hippocampus are expanded and diversified in APP and Pilo mice (vs. respective controls). Domains of interest in both disease contexts involved neuronal excitability and neurotransmission, neurogenesis, chromatin remodeling, and cellular stress and neuroinflammation. To assess the gene targets bound by ΔFosB regardless of seizure etiology, we focused on 442 genes with significant ΔFosB binding in both APP and Pilo mice (vs. respective controls). Functional analyses identified pathways that regulate membrane potential, glutamatergic signaling, calcium homeostasis, complement activation, neuron-glia population maintenance, and chromatin dynamics. RNA-sequencing and qPCR measurements in independent mice detected altered expression of several ΔFosB targets shared in APP and Pilo mice. Our findings indicate that seizure-induced ΔFosB can bind genes in patterns that depend on seizure etiology, but can bind other genes regardless of seizure etiology. Understanding the factors that underlie these differences, such as chromatin accessibility and/or abundance of co-factors, could reveal novel insights into the control of gene expression in disorders with recurrent seizures.
PMCID:7268090
PMID: 32536852
ISSN: 1662-4548
CID: 4484462